Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Publication year range
1.
Am J Clin Nutr ; 118(1): 303-313, 2023 07.
Article in English | MEDLINE | ID: mdl-37217097

ABSTRACT

BACKGROUND: Prenatal exposure to antiseizure medication (ASM) may lead to low plasma folate concentrations and is associated with impaired neurodevelopment. OBJECTIVES: To examine whether maternal genetic liability to folate deficiency interacts with ASM-associated risk of language impairment and autistic traits in children of women with epilepsy. METHODS: We included children of women with and without epilepsy and with available genetic data enrolled in the Norwegian Mother, Father, and Child Cohort Study. Information on ASM use, folic acid supplement use and dose, dietary folate intake, child autistic traits, and child language impairment was obtained from parent-reported questionnaires. Using logistic regression, we examined the interaction between prenatal ASM exposure and maternal genetic liability to folate deficiency expressed as polygenic risk score of low folate concentrations or maternal rs1801133 genotype (CC or CT/TT) on risk of language impairment or autistic traits. RESULTS: We included 96 children of women with ASM-treated epilepsy, 131 children of women with ASM-untreated epilepsy, and 37,249 children of women without epilepsy. The polygenic risk score of low folate concentrations did not interact with the ASM-associated risk of language impairment or autistic traits in ASM-exposed children of women with epilepsy compared with ASM-unexposed children aged 1.5-8 y. ASM-exposed children had increased risk of adverse neurodevelopment regardless of maternal rs1801133 genotype {adjusted odds ratio [aOR] for language impairment aged 8 y was 2.88 [95% confidence interval (CI): 1.00, 8.26] if CC and aOR 2.88 [95% CI: 1.10, 7.53] if CT/TT genotypes}. In children of women without epilepsy aged 3 y, those with maternal rs1801133 CT/TT compared with CC genotype had increased risk of language impairment (aOR: 1.18; 95% CI: 1.05, 1.34). CONCLUSIONS: In this cohort of pregnant women reporting widespread use of folic acid supplements, maternal genetic liability to folate deficiency did not significantly influence the ASM-associated risk of impaired neurodevelopment.


Subject(s)
Autistic Disorder , Epilepsy , Folic Acid Deficiency , Language Development Disorders , Prenatal Exposure Delayed Effects , Humans , Child , Female , Pregnancy , Cohort Studies , Autistic Disorder/genetics , Autistic Disorder/drug therapy , Folic Acid , Epilepsy/drug therapy , Epilepsy/genetics , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/drug therapy , Language Development Disorders/drug therapy
2.
Andrology ; 11(5): 927-942, 2023 07.
Article in English | MEDLINE | ID: mdl-36697378

ABSTRACT

BACKGROUND: DNA methylation (DNAme) erasure and reacquisition occur during prenatal male germ cell development; some further remodeling takes place after birth during spermatogenesis. Environmental insults during germline epigenetic reprogramming may affect DNAme, presenting a potential mechanism for transmission of environmental exposures across multiple generations. OBJECTIVES: We investigated how germ cell DNAme is impacted by lifetime exposures to diets containing either low or high, clinically relevant, levels of the methyl donor folic acid and whether resulting DNAme alterations were inherited in germ cells of male offspring of subsequent generations. MATERIALS AND METHODS: Female mice were placed on a control (FCD), 7-fold folic acid deficient (7FD) or 10- to 20-fold supplemented (10FS and 20FS) diet before and during pregnancy. Resulting F1 litters were weaned on the respective diets. F2 and F3 males received control diets. Genome-wide DNAme at cytosines (within CpG sites) was assessed in F1 spermatogonia, and in F1, F2 and F3 sperm. RESULTS: In F1 germ cells, a greater number of differentially methylated cytosines (DMCs) were observed in spermatogonia as compared with F1 sperm for all folic acid diets. DMCs were lower in number in F2 versus F1 sperm, while an unexpected increase was found in F3 sperm. DMCs were predominantly hypomethylated, with genes in neurodevelopmental pathways commonly affected in F1, F2 and F3 male germ cells. While no DMCs were found to be significantly inherited inter- or transgenerationally, we observed over-representation of repetitive elements, particularly young long interspersed nuclear elements (LINEs). DISCUSSION AND CONCLUSION: These results suggest that the prenatal window is the time most susceptible to folate-induced alterations in sperm DNAme in male germ cells. Altered methylation of specific sites in F1 germ cells was not present in later generations. However, the presence of DNAme perturbations in the sperm of males of the F2 and F3 generations suggests that epigenetic inheritance mechanisms other than DNAme may have been impacted by the folate diet exposure of F1 germ cells.


Subject(s)
DNA Methylation , Folic Acid Deficiency , Pregnancy , Male , Female , Mice , Animals , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Semen/metabolism , Epigenesis, Genetic , Spermatozoa/metabolism , Folic Acid/metabolism , Dietary Supplements , Spermatogonia/metabolism , DNA/metabolism
3.
Mol Nutr Food Res ; 66(3): e2100789, 2022 02.
Article in English | MEDLINE | ID: mdl-34850562

ABSTRACT

SCOPE: Persistent DNA methylation changes may mediate effects of early-life exposures on later-life health. Human lifespan is challenging for prospective studies, therefore data from longitudinal studies are limited. Projecting data from mouse models of early-life exposure to human studies offers a tool to address this challenge. METHODS AND RESULTS: C57BL/6J mice were fed low/normal folate diets before and during pregnancy and lactation. Genome-wide promoter methylation was measured in male offspring livers at 17.5 days gestation and 28 weeks. Eight promoters were concurrently hypermethylated by folate depletion in fetuses and adults (>1.10 fold-change; p < 0.05). Processes/pathways potentially influenced by global changes, and function of these eight genes, suggest neurocognitive effects. Human observational and randomized controlled trial data were interrogated for translation. Methylation at birth was inversely associated with maternal plasma folate in six genes (-1.15% to -0.16% per nmol L-1 ; p < 0.05), while maternal folic acid supplementation was associated with differential methylation of four genes in adulthood. Three CpGs were persistently hypermethylated with lower maternal folate (p = 0.04). CONCLUSION: Some persistent folate-induced methylation changes in mice are mirrored in humans. This demonstrates utility of mouse data in identifying human loci for interrogation as biomarkers of later-life health.


Subject(s)
DNA Methylation , Folic Acid Deficiency , Adult , Animals , Female , Folic Acid/pharmacology , Folic Acid Deficiency/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prospective Studies
4.
PLoS One ; 16(12): e0260860, 2021.
Article in English | MEDLINE | ID: mdl-34855899

ABSTRACT

BACKGROUND: Anthropogenic air pollution has been implicated in aberrant changes of DNA methylation and homocysteine increase (>15µM/L). Folate (<3 ng/mL) and vitamin B12 (<220 pg/mL) deficiencies also reduce global DNA methylation via homocysteine increase. Although B-vitamin supplements can attenuate epigenetic effects of air pollution but such understanding in population-specific studies are lacking. Hence, the present study aims to understand the role of air pollution, homocysteine, and nutritional deficiencies on methylation. METHODS: We examined cross-sectionally, homocysteine, folate, vitamin B12 (chemiluminescence) and global DNA methylation (colorimetric ELISA Assay) among 274 and 270 individuals from low- and high- polluted areas, respectively, from a single Mendelian population. Global DNA methylation results were obtained on 254 and 258 samples from low- and high- polluted areas, respectively. RESULTS: Significant decline in median global DNA methylation was seen as a result of air pollution [high-0.84 (0.37-1.97) vs. low-0.96 (0.45-2.75), p = 0.01]. High homocysteine in combination with air pollution significantly reduced global DNA methylation [high-0.71 (0.34-1.90) vs. low-0.93 (0.45-3.00), p = 0.003]. Folate deficient individuals in high polluted areas [high-0.70 (0.37-1.29) vs. low-1.21 (0.45-3.65)] showed significantly reduced global methylation levels (p = 0.007). In low polluted areas, despite folate deficiency, if normal vitamin B12 levels were maintained, global DNA methylation levels improved significantly [2.03 (0.60-5.24), p = 0.007]. Conversely, in high polluted areas despite vitamin B12 deficiency, if normal folate status was maintained, global DNA methylation status improved significantly [0.91 (0.36-1.63)] compared to vitamin B12 normal individuals [0.54 (0.26-1.13), p = 0.04]. CONCLUSIONS: High homocysteine may aggravate the effects of air pollution on DNA methylation. Vitamin B12 in low-polluted and folate in high-polluted areas may be strong determinants for changes in DNA methylation levels. The effect of air pollution on methylation levels may be reduced through inclusion of dietary or supplemented B-vitamins. This may serve as public level approach in natural settings to prevent metabolic adversities at community level.


Subject(s)
Air Pollution/analysis , DNA Methylation , Folic Acid Deficiency/epidemiology , Homocysteine/blood , Hyperhomocysteinemia/epidemiology , Vitamin B 12 Deficiency/epidemiology , Adult , Aged , Air Pollution/adverse effects , Cross-Sectional Studies , Female , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , Humans , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/genetics , India/epidemiology , Male , Middle Aged , Vitamin B 12 Deficiency/blood , Vitamin B 12 Deficiency/genetics
5.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34546337

ABSTRACT

Hereditary spastic paraplegias are heterogeneous neurodegenerative disorders. Understanding of their pathogenic mechanisms remains sparse, and therapeutic options are lacking. We characterized a mouse model lacking the Cyp2u1 gene, loss of which is known to be involved in a complex form of these diseases in humans. We showed that this model partially recapitulated the clinical and biochemical phenotypes of patients. Using electron microscopy, lipidomic, and proteomic studies, we identified vitamin B2 as a substrate of the CYP2U1 enzyme, as well as coenzyme Q, neopterin, and IFN-α levels as putative biomarkers in mice and fluids obtained from the largest series of CYP2U1-mutated patients reported so far. We also confirmed brain calcifications as a potential biomarker in patients. Our results suggest that CYP2U1 deficiency disrupts mitochondrial function and impacts proper neurodevelopment, which could be prevented by folate supplementation in our mouse model, followed by a neurodegenerative process altering multiple neuronal and extraneuronal tissues.


Subject(s)
Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Folic Acid/pharmacology , Animals , Biomarkers/metabolism , Brain/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Phenotype , Proteomics/methods
6.
Nat Genet ; 53(8): 1233-1242, 2021 08.
Article in English | MEDLINE | ID: mdl-34326545

ABSTRACT

The agouti viable yellow (Avy) allele is an insertional mutation in the mouse genome caused by a variably methylated intracisternal A particle (VM-IAP) retrotransposon. Avy expressivity is sensitive to a range of early-life chemical exposures and nutritional interventions, suggesting that environmental perturbations can have long-lasting effects on the methylome. However, the extent to which VM-IAP elements are environmentally labile with phenotypic implications is unknown. Using a recently identified repertoire of VM-IAPs, we assessed the epigenetic effects of different environmental contexts. A longitudinal aging analysis indicated that VM-IAPs are stable across the murine lifespan, with only small increases in DNA methylation detected for a subset of loci. No significant effects were observed after maternal exposure to the endocrine disruptor bisphenol A, an obesogenic diet or methyl donor supplementation. A genetic mouse model of abnormal folate metabolism exhibited shifted VM-IAP methylation levels and altered VM-IAP-associated gene expression, yet these effects are likely largely driven by differential targeting by polymorphic KRAB zinc finger proteins. We conclude that epigenetic variability at retrotransposons is not predictive of environmental susceptibility.


Subject(s)
DNA Methylation , Endocrine Disruptors/toxicity , Obesity/genetics , Retroelements , Animals , Benzhydryl Compounds/toxicity , DNA Methylation/drug effects , Diet/adverse effects , Epigenesis, Genetic , Female , Ferredoxin-NADP Reductase/genetics , Folic Acid/genetics , Folic Acid/metabolism , Folic Acid Deficiency/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Obesity/etiology , Phenols/toxicity , Pregnancy , Prenatal Exposure Delayed Effects
7.
Mol Reprod Dev ; 88(6): 437-458, 2021 06.
Article in English | MEDLINE | ID: mdl-34008284

ABSTRACT

Genomic imprinting is important for mammalian development and its dysregulation can cause various developmental defects and diseases. The study evaluated the effects of different dietary combinations of folic acid and B12 on epigenetic regulation of IGF2R and KCNQ1OT1 ncRNA in C57BL/6 mice model. Female mice were fed diets with nine combinations of folic acid and B12 for 4 weeks. They were mated and off-springs born (F1) were continued on the same diet for 6 weeks postweaning and were allowed to mate. The placenta and fetal (F2) tissues were collected at day 20 of gestation. Dietary deficiency of folate (BNFD and BOFD) and B12 (BDFN) with either state of other vitamin or combined deficiency of both vitamins (BDFD) in comparison to BNFN, were overall responsible for reduced expression of IGF2R in the placenta (F1) and the fetal liver (F2) whereas a combination of folate deficiency with different levels of B12 revealed sex-specific differences in kidney and brain. The alterations in the expression of IGF2R caused by folate-deficient conditions (BNFD and BOFD) and both deficient condition (BDFD) was found to be associated with an increase in suppressive histone modifications. Over-supplementation of either folate or B12 or both vitamins in comparison to BNFN, led to increase in expression of IGF2R and KCNQ1OT1 in the placenta and fetal tissues. The increase in the expression of IGF2R caused by folate over-supplementation (BNFO) was associated with decreased DNA methylation in fetal tissues. KCNQ1OT1 noncoding RNA (ncRNA), however, showed upregulation under deficient conditions of folate and B12 only in female fetal tissues which correlated well with hypomethylation observed under these conditions. An epigenetic reprograming of IGF2R and KCNQ1OT1 ncRNA in the offspring was evident upon different dietary combinations of folic acid and B12 in the mice.


Subject(s)
Diet , Epigenesis, Genetic/drug effects , Fetus/drug effects , Folic Acid/pharmacology , Gene Expression Regulation, Developmental/drug effects , Placenta/drug effects , RNA, Long Noncoding/genetics , Receptor, IGF Type 2/genetics , Vitamin B 12/pharmacology , Animals , Body Weight/drug effects , Brain/embryology , Brain/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Female , Fetus/metabolism , Folic Acid/administration & dosage , Folic Acid/blood , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Genomic Imprinting , Homocysteine/blood , Kidney/embryology , Kidney/metabolism , Liver/embryology , Liver/metabolism , Male , Mice , Placenta/metabolism , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , RNA, Long Noncoding/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, IGF Type 2/metabolism , Vitamin B 12/administration & dosage , Vitamin B 12/blood , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/metabolism
8.
J Nutr ; 151(4): 800-809, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33693772

ABSTRACT

BACKGROUND: While cancer is common, its incidence varies widely by tissue. These differences are attributable to variable risk factors, such as environmental exposure, genetic inheritance, and lifetime number of stem cell divisions in a tissue. Folate deficiency is generally associated with increased risk for colorectal cancer (CRC) and acute lymphocytic leukemia (ALL). Conversely, high folic acid (FA) intake has also been associated with higher CRC risk. OBJECTIVE: Our objective was to compare the effect of folate intake on mutant frequency (MF) and types of mutations in the colon and bone marrow of mice. METHODS: Five-week-old MutaMouse male mice were fed a deficient (0 mg FA/kg), control (2 mg FA/kg), or supplemented (8 mg FA/kg) diet for 20 wk. Tissue MF was assessed using the lacZ mutant assay and comparisons made by 2-factor ANOVA. LacZ mutant plaques were sequenced using next-generation sequencing, and diet-specific mutation profiles within each tissue were compared by Fisher's exact test. RESULTS: In the colon, the MF was 1.5-fold and 1.3-fold higher in mice fed the supplemented diet compared with mice fed the control (P = 0.001) and deficient (P = 0.008) diets, respectively. This contrasted with the bone marrow MF in the same mice where the MF was 1.7-fold and 1.6-fold higher in mice fed the deficient diet compared with mice fed the control (P = 0.02) and supplemented (P = 0.03) diets, respectively. Mutation profiles and signatures (mutation context) were tissue-specific. CONCLUSIONS: Our data indicate that dietary folate intake affects mutagenesis in a tissue- and dose-specific manner in mice. Mutation profiles were generally tissue- but not dose-specific, suggesting that altered cellular folate status appears to interact with endogenous mutagenic mechanisms in each tissue to create a permissive context in which specific mutation types accumulate. These data illuminate potential mechanisms underpinning differences in observed associations between folate intake/status and cancer.


Subject(s)
Folic Acid/administration & dosage , Mutation Rate , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Colon/drug effects , Colon/metabolism , Dose-Response Relationship, Drug , Folic Acid/adverse effects , Folic Acid/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , High-Throughput Nucleotide Sequencing , Lac Operon/drug effects , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis , Organ Specificity
9.
Ophthalmologe ; 118(4): 383-390, 2021 Apr.
Article in German | MEDLINE | ID: mdl-32632495

ABSTRACT

Cerebral folate deficiency (CFD) results in neurological alterations and a massive degeneration of the choroid/retina if left untreated, which limit the visual field and visual acuity. This article reports the case of a female patient with CFD, who developed autistic personal characteristics prior to reaching school age and first started to speak at the age of 3 years. At the age of 6 years she was presented because of unclear reduced visual acuity in the right eye. At that time mild bilateral peripheral chorioretinal atrophy was present, which subsequently became more pronounced. Additionally, a centrally emphasized chorioretinal atrophy further developed. Visual acuity of both eyes progressively deteriorated until stagnating at 0.1 at the age of 14 years. The causal assignment of the findings of the patient was not possible for many years. Choroideremia was excluded by molecular genetic testing (CHM gene with no mutations) and gyrate atrophy was ruled out by a normal ornithine level. The existence of a mitochondrial disease was almost completely excluded by exome sequencing. After the onset of further nonocular symptoms, e.g. neuromuscular disorders, electroencephalograph (EEG) alterations and autistic disorder, intensified laboratory diagnostics were performed in the treating pediatric hospital. Finally, an extremely low level of the folic acid metabolite 5­methyltetrahydrofolate was detected in the cerebrospinal fluid (CSF) leading to the diagnosis of CFD. High-dose substitution treatment with folic acid was subsequently initiated. After excluding the presence of a pathogenic mutation of the FOLR1 gene for the cerebral folate receptor 1, a high titer blocking autoantibody against cerebral folate receptor 1 was detected as the cause.


Subject(s)
Folic Acid Deficiency , Retinal Degeneration , Adolescent , Atrophy , Child , Child, Preschool , Female , Folate Receptor 1/genetics , Folic Acid , Folic Acid Deficiency/diagnosis , Folic Acid Deficiency/drug therapy , Folic Acid Deficiency/genetics , Humans
10.
Cereb Cortex ; 31(1): 635-649, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32995858

ABSTRACT

Folate is an essential micronutrient required for both cellular proliferation through de novo nucleotide synthesis and epigenetic regulation of gene expression through methylation. This dual requirement places a particular demand on folate availability during pregnancy when both rapid cell generation and programmed differentiation of maternal, extraembryonic, and embryonic/fetal tissues are required. Accordingly, prenatal neurodevelopment is particularly susceptible to folate deficiency, which can predispose to neural tube defects, or when effective transport into the brain is impaired, cerebral folate deficiency. Consequently, adequate folate consumption, in the form of folic acid (FA) fortification and supplement use, is widely recommended and has led to a substantial increase in the amount of FA intake during pregnancy in some populations. Here, we show that either maternal folate deficiency or FA excess in mice results in disruptions in folate metabolism of the offspring, suggesting diversion of the folate cycle from methylation to DNA synthesis. Paradoxically, either intervention causes comparable neurodevelopmental changes by delaying prenatal cerebral cortical neurogenesis in favor of late-born neurons. These cytoarchitectural and biochemical alterations are accompanied by behavioral abnormalities in FA test groups compared with controls. Our findings point to overlooked potential neurodevelopmental risks associated with excessively high levels of prenatal FA intake.


Subject(s)
Behavior, Animal/drug effects , Epigenesis, Genetic/drug effects , Folic Acid/pharmacology , Pregnancy/drug effects , Animals , DNA Methylation/drug effects , Dietary Supplements/adverse effects , Female , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Mice, Inbred C57BL
11.
Nutrients ; 12(9)2020 Sep 19.
Article in English | MEDLINE | ID: mdl-32961717

ABSTRACT

Vitamins B9 (folate) and B12 are essential water-soluble vitamins that play a crucial role in the maintenance of one-carbon metabolism: a set of interconnected biochemical pathways driven by folate and methionine to generate methyl groups for use in DNA synthesis, amino acid homeostasis, antioxidant generation, and epigenetic regulation. Dietary deficiencies in B9 and B12, or genetic polymorphisms that influence the activity of enzymes involved in the folate or methionine cycles, are known to cause developmental defects, impair cognitive function, or block normal blood production. Nutritional deficiencies have historically been treated with dietary supplementation or high-dose parenteral administration that can reverse symptoms in the majority of cases. Elevated levels of these vitamins have more recently been shown to correlate with immune dysfunction, cancer, and increased mortality. Therapies that specifically target one-carbon metabolism are therefore currently being explored for the treatment of immune disorders and cancer. In this review, we will highlight recent studies aimed at elucidating the role of folate, B12, and methionine in one-carbon metabolism during normal cellular processes and in the context of disease progression.


Subject(s)
Folic Acid Deficiency/prevention & control , Folic Acid/pharmacology , One-Carbon Group Transferases/metabolism , Vitamin B 12 Deficiency/prevention & control , Vitamin B 12/pharmacology , Folic Acid Deficiency/genetics , Humans , Polymorphism, Genetic , Vitamin B 12 Deficiency/genetics
12.
Clin Nutr ; 39(12): 3771-3778, 2020 12.
Article in English | MEDLINE | ID: mdl-32334879

ABSTRACT

BACKGROUND & AIMS: The 4977-bp mitochondrial deletion (mtDNA4977 deletion), as a hallmark of mitochondrial oxidative damage, may play an important role in coronary artery disease (CAD), but its interaction with folate deficiency among diabetic patients is largely unknown. We aimed to explore the joint association of leukocyte mtDNA4977 deletion and serum folate status with obstructive CAD in Chinese adults with type 2 diabetes. METHODS: We cross-sectionally analyzed the angiographic data of 2017 diabetic patients without B-vitamin supplementation. Of the 2017 participants, 756 who received percutaneous coronary intervention (PCI) completed prospective follow-up of one year. In vitro, we explored the mediation effects of mitochondrial reactive oxygen species (mtROS) in folic acid (FA)-deficient human aortic smooth muscle cells (HASMCs) under hyperglycemic conditions. RESULTS: Cross-sectionally, the multivariate odds ratios (ORs) for obstructive CAD were 1.41 (95% CI: 1.29-1.55) for greater mtDNA4977 deletion, and 1.15 (95% CI: 1.05-1.25) for lower folate levels. Particularly, the combination of high mtDNA4977 deletion (top tertile) and folate deficiency (serum folate < 6 ng/mL) was associated with more than 2-fold increased odds of having obstructive CAD and higher degrees of coronary stenosis. Prospectively, the hazard ratio for all-cause death at 1-year after PCI was up to 2.37 (95% CI: 1.21-4.63) for folate-deficient participants in the top tertile of mtDNA4977 deletion. In HASMCs, the adverse effects of FA deficiency were aggravated by induction of mtROS, and attenuated by scavenging of mtROS. CONCLUSIONS: The risk of obstructive CAD may be greatly increased by the interaction between greater mtDNA4977 deletion and folate deficiency among diabetic patients.


Subject(s)
Coronary Artery Disease/complications , DNA, Mitochondrial/blood , Diabetes Mellitus, Type 2/complications , Folic Acid Deficiency/complications , Folic Acid/blood , Aged , China , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Female , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , Humans , Leukocytes , Male , Middle Aged , Odds Ratio , Percutaneous Coronary Intervention , Proportional Hazards Models , Prospective Studies
13.
Reprod Sci ; 27(4): 963-976, 2020 04.
Article in English | MEDLINE | ID: mdl-32124397

ABSTRACT

The clinical significance of periconceptional folic acid supplementation (FAS) in the prevention of neonatal neural tube defects (NTDs) has been recognized for decades. Epidemiological data and experimental findings have consistently been indicating an association between folate deficiency in the first trimester of pregnancy and poor fetal development as well as offspring health (i.e., NTDs, isolated orofacial clefts, neurodevelopmental disorders). Moreover, compelling evidence has suggested adverse effects of folate overload during perinatal period on offspring health (i.e., immune diseases, autism, lipid disorders). In addition to several single-nucleotide polymorphisms (SNPs) in genes related to folate one-carbon metabolism (FOCM), folate concentrations in maternal serum/plasma/red blood cells must be considered when counseling FAS. Epigenetic information encoded by 5-methylcytosines (5mC) plays a critical role in fetal development and offspring health. S-adenosylmethionine (SAM), a methyl donor for 5mC, could be derived from FOCM. As such, folic acid plays a double-edged sword role in offspring health via mediating DNA methylation. However, the underlying epigenetic mechanism is still largely unclear. In this review, we summarized the link across DNA methylation, maternal FAS, and offspring health to provide more evidence for clinical guidance in terms of precise FAS dosage and time point. Future studies are, therefore, required to set up the reference intervals of folate concentrations at different trimesters of pregnancy for different populations and to clarify the epigenetic mechanism for specific offspring diseases.


Subject(s)
DNA Methylation/drug effects , Epigenesis, Genetic , Folic Acid/administration & dosage , Maternal Nutritional Physiological Phenomena/genetics , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Animals , Dietary Supplements , Female , Fetal Development/drug effects , Folic Acid/adverse effects , Folic Acid Deficiency/genetics , Humans , Maternal Nutritional Physiological Phenomena/drug effects , Pregnancy
14.
Biochimie ; 173: 27-32, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32061804

ABSTRACT

Neural tube defects (NTDs) are a broad class of congenital birth defects that result from the failure of neural tube closure during neurulation. Folic acid supplementation has been shown to prevent the occurrence of NTDs by as much as 70% in some human populations, and folate deficiency in a pregnant woman is associated with increased risk for having an NTD affected infant. Thus, folate transport-related genes and genes involved in the subsequent folate-mediated one-carbon metabolic pathway have long been considered primary candidates to study the genetic etiology of human NTDs. Herein, we review the genes involved in folate transport and one-carbon metabolism thus far identified as contributing variants that influence human NTD risk, and place these findings in the context of our evolving understanding of the complex genetic architecture underlying these defects.


Subject(s)
Biological Transport/genetics , Folic Acid Deficiency/genetics , Folic Acid/metabolism , Metabolic Networks and Pathways/genetics , Neural Tube Defects/genetics , Female , Humans , Pregnancy
15.
Br J Nutr ; 123(3): 264-272, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31656209

ABSTRACT

The present study aimed at analysing how dietary folic acid (FA) and Fe deficiency, followed by supplementation with these nutrients, affects the expression of folate and Fe transporters in the duodenum, as well as FA and Fe status. After a deficiency period, Wistar rats were randomised to a group fed with a diet deficient in FA and supplemented with Fe (DFE), a diet deficient in Fe and supplemented with FA, a diet supplemented with Fe and FA (FEFOL), a diet deficient in Fe and FA (D) or a control diet (C). Tissue collection was performed after 2, 10 or 21 d of these diets. Group D had higher Slc11a2 mRNA levels than the DFE group at every time point and there were differences in mRNA levels of Slc46a1 between the DFE and the FEFOL groups at the third time point, but we observed no differences in protein levels between the groups. The DFE and D groups not only had lower serum folate concentrations at every time point but also had the highest homocysteine concentrations. Total Fe binding capacity concentrations were the lowest in the DFE group at the first time point and in the DFE and the FEFOL groups at the final time point. Simultaneous supplementation with FA and Fe resulted in significantly higher Hb concentrations than did supplementation with these nutrients alone. Our findings indicate that dietary FA and Fe deficiency, and subsequent supplementation with these nutrients, affects transcription but not the protein levels of FA and Fe transporters in the duodenum.


Subject(s)
Anemia, Iron-Deficiency/genetics , Dietary Supplements , Folic Acid Deficiency/genetics , Folic Acid/administration & dosage , Iron/administration & dosage , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/etiology , Animals , Cation Transport Proteins/metabolism , Diet/adverse effects , Duodenum/metabolism , Folic Acid/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/etiology , Iron/blood , Nutritional Status , Proton-Coupled Folate Transporter/metabolism , Rats , Rats, Wistar , Transcription, Genetic/drug effects
16.
Article in English | MEDLINE | ID: mdl-31421742

ABSTRACT

Intake of folate (vitamin B9) is strongly inversely linked with human cancer risk, particularly colon cancer. In general, people with the highest dietary intake of folate or with high blood folate levels are at a reduced risk (approx. 25%) of developing colon cancer. Folate acts in normal cellular metabolism to maintain genomic stability through the provision of nucleotides for DNA replication and DNA repair and by regulating DNA methylation and gene expression. Folate deficiency can accelerate carcinogenesis by inducing misincorporation of uracil into DNA, by increasing DNA strand breakage, by inhibiting DNA base excision repair capacity and by inducing DNA hypomethylation and consequently aberrant gene and protein expression. Conversely, increasing folate intake may improve genomic stability. This review describes key applications of single cell gel electrophoresis (the comet assay) in assessing genomic instability (misincorporated uracil, DNA single strand breakage and DNA repair capacity) in response to folate status (deficient or supplemented) in human cells in vitro, in rodent models and in human case-control and intervention studies. It highlights an adaptation of the SCGE comet assay for measuring genome-wide and gene-specific DNA methylation in human cells and colon tissue.


Subject(s)
Biological Monitoring/methods , Colonic Neoplasms/genetics , Comet Assay/methods , Folic Acid/pharmacology , Genomic Instability , Single-Cell Analysis/methods , Cell Line , Colonic Neoplasms/epidemiology , Colonic Neoplasms/prevention & control , DNA Breaks , DNA Methylation , DNA Repair , DNA Replication , Folic Acid/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , Genomic Instability/drug effects , Genomic Instability/genetics , Genotype , Homocystinuria/blood , Homocystinuria/genetics , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/blood , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/physiology , Muscle Spasticity/blood , Muscle Spasticity/genetics , Psychotic Disorders/blood , Psychotic Disorders/genetics , Risk , Uracil/metabolism
17.
Int J Mol Sci ; 20(5)2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30836646

ABSTRACT

Supplementation of micronutrients like folate is a double-edged sword in terms of their ambivalent role in cell metabolism. Although several epidemiological studies support a protective role of folate in carcinogenesis, there are also data arguing for an opposite effect. To address this issue in the context of human papillomavirus (HPV)-induced transformation, the molecular events of different folate availability on human keratinocytes immortalized by HPV16 E6 and E7 oncoproteins were examined. Several sublines were established: Control (4.5 µM folate), folate deficient (0.002 µM folate), and repleted cells (4.5 µM folate). Cells were analyzed in terms of oncogene expression, DNA damage and repair, karyotype changes, whole-genome sequencing, and transcriptomics. Here we show that folate depletion irreversibly induces DNA damage, impairment of DNA repair fidelity, and unique chromosomal alterations. Repleted cells additionally underwent growth advantage and enhanced clonogenicity, while the above mentioned impaired molecular properties became even more pronounced. Overall, it appears that a period of folate deficiency followed by repletion can shape immortalized cells toward an anomalous phenotype, thereby potentially contributing to carcinogenesis. These observations should elicit questions and inquiries for broader additional studies regarding folate fortification programs, especially in developing countries with micronutrient deficiencies and high HPV prevalence.


Subject(s)
Folic Acid Deficiency/genetics , Human papillomavirus 16/genetics , Papillomavirus Infections/genetics , Transcription, Genetic , Carcinogenesis/genetics , DNA Damage/ethics , DNA Repair/genetics , Folic Acid/genetics , Folic Acid Deficiency/pathology , Folic Acid Deficiency/virology , Genomics , Human papillomavirus 16/pathogenicity , Humans , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , Repressor Proteins/genetics
18.
J Neurol Sci ; 396: 112-118, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30448717

ABSTRACT

OBJECTIVE: To describe the phenotype and the response to folinic acid supplementation of cerebral folate deficiency (CFD) in adults, a disorder diagnosed on low 5-methyltetrahydro-folate (5MTHF) in cerebrospinal fluid (CSF), which can correspond to a inherited disorder of folate metabolism (IDFM) or to a metabolic consequence of various neurological diseases. METHODS: We conducted a retrospective study on 224 adult patients with neurological symptoms who had a 5MTHF CSF dosage, collecting their neurologic and neuroimaging data. RESULTS: 69 patients had CFD (CSF 5MTHF level < 41 nmol/L), 25 of them had severe CFD (sCFD; ≤25 nmol/L) with adult onset neurological symptoms in 41%. 56% of sCFD patients had an underlying identified neurologic disorder, mainly mitochondrial diseases, hepatic encephalopathy and primary brain calcifications (no identified IDFM), the others were classified as undiagnosed. sCFD patients presented most frequently pyramidal syndrome (75%), movement disorders (56%), cerebellar syndrome (50%) and intellectual disability (46%). MRI findings mostly showed white matter abnormalities (WMA; 32%) and calcifications (12%), and were normal in 23%. The clinico-radiological phenotype of sCFD patients was not clearly different from non CFD patients in terms of manifestations frequency. However, their neurological picture was more complex with a higher number of combined neurological symptoms (4.7±1.6 vs 3.4±1.7, p = .01). In Magnetic Resonance Spectroscopy (MRS), Choline/Creatine (Cho/Cr) ratio was lower in sCFD patients (n = 7) compared to non-CFD patients (n = 73) (p = .005), with good sensitivity (71%) and excellent specificity (92%). Among twenty-one CFD patients treated with folinic acid, nine had a sustained improvement, all with sCFD but one (50% of sCFD patients improved). In two undiagnosed patients with extremely low 5MTHF CSF values, MRI WMA and low Cho/Cr ratios, folinic acid treatment leaded to a dramatic clinical and radiological improvement. CONCLUSION: CSF 5MTHF dosage should be considered in patients with mitochondrial diseases, primary brain calcifications and unexplained complex neurological disorders especially if associated with WMA, since folinic acid supplementation in patients with sCFD is frequently efficient.


Subject(s)
Cerebellar Diseases/complications , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Mutation/genetics , Proteins/genetics , Adolescent , Adult , Aged , Calcinosis/diagnostic imaging , Calcinosis/etiology , Calcinosis/genetics , Cerebellar Diseases/cerebrospinal fluid , Cerebellar Diseases/diagnostic imaging , Cerebellar Diseases/genetics , Child , Child, Preschool , Female , Folic Acid/cerebrospinal fluid , Folic Acid Deficiency/cerebrospinal fluid , Folic Acid Deficiency/diagnostic imaging , Follow-Up Studies , Humans , Infant , Infant, Newborn , Intellectual Disability , Magnetic Resonance Imaging , Male , Middle Aged , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Diseases/etiology , Retrospective Studies , Tetrahydrofolates/cerebrospinal fluid , White Matter/diagnostic imaging , White Matter/pathology , Young Adult
19.
J Clin Neurosci ; 59: 341-344, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30420205

ABSTRACT

INTRODUCTION: Folate is essential for production of DNA, neurotransmitters and myelin and regulation of genetic activity. A specific transporter protein is required to transport folate from blood to CSF. Various inherited brain-specific folate transport defects have been recognized due to mutation in Folate Receptor alpha (FOLR1). FOLR1 mutation is one of the vitamin responsive encephalopathies and is inherited as an autosomal recessive condition. It has a wide spectrum of phenotype, commonly presenting as epileptic encephalopathy. Less frequently the condition may manifest with subtle hypotonia, movement disorder as tremors, ataxia or intellectual disability and autistic spectrum disorder. We present a case of folate transporter deficiency with non-epileptic manifestations, presenting with tremors, speech delay and stable white matter changes in MRI brain. OBJECTIVE: We present a case of Folate transporter defect with Non-epileptic presentation. CONCLUSION: Folate transporter deficiency has a wide range of presenting symptoms. Presentation with slowly progressive atypical symptoms, stable white matter changes in brain MRI that does not fit a specific diagnosis, should raise a high suspicion of FOLR1 mutation, even in absence of seizures. Since folate transporter deficiency is a treatable neurodegenerative disorder, early diagnosis and supplementation with folinic acid is vital.


Subject(s)
Folic Acid Deficiency/pathology , Malabsorption Syndromes/pathology , Phenotype , Brain/metabolism , Child , Female , Folate Receptor 1/genetics , Folic Acid Deficiency/diagnostic imaging , Folic Acid Deficiency/genetics , Humans , Malabsorption Syndromes/diagnostic imaging , Malabsorption Syndromes/genetics , Mutation
20.
Microbiology (Reading) ; 164(7): 982-991, 2018 07.
Article in English | MEDLINE | ID: mdl-29799386

ABSTRACT

Dihydrofolate reductase (DHFR) and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase/IMP cyclohydrolase (PurH) play key roles in maintaining folate pools in cells, and are targets of antimicrobial and anticancer drugs. While the activities of bacterial DHFR and PurH on their classical substrates (DHF and 10-CHO-THF, respectively) are known, their activities and kinetic properties of utilisation of 10-CHO-DHF are unknown. We have determined the kinetic properties (kcat/Km) of conversion of 10-CHO-DHF to 10-CHO-THF by DHFR, and to DHF by PurH. We show that DHFR utilises 10-CHO-DHF about one third as efficiently as it utilises DHF. The 10-CHO-DHF is also utilised (as a formyl group donor) by PurH albeit slightly less efficiently than 10-CHO-THF. The utilisation of 10-CHO-DHF by DHFR is ~50 fold more efficient than its utilisation by PurH. A folate deficient Escherichia coli (∆pabA) grows well when supplemented with adenine, glycine, thymine and methionine, the metabolites that arise from the one-carbon metabolic pathway. Notably, when the ∆pabA strain harboured a folate transporter, it grew in the presence of 10-CHO-DHF alone, suggesting that it (10-CHO-DHF) can enter one-carbon metabolic pathway to provide the required metabolites. Thus, our studies reveal that both DHFR and PurH could utilise 10-CHO-DHF for folate homeostasis in E. coli.


Subject(s)
Escherichia coli/metabolism , Folic Acid/analogs & derivatives , Nucleotide Deaminases/metabolism , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , 4-Aminobenzoic Acid , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/growth & development , Folic Acid/metabolism , Folic Acid Deficiency/genetics , Homeostasis , Kinetics , Metabolic Networks and Pathways , Nucleotide Deaminases/genetics , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/genetics , Tetrahydrofolate Dehydrogenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL